Language selection

Search

Patent 2476427 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 2476427
(54) English Title: PARTIAL PEPTIDE MIMETICS AND METHODS
(54) French Title: MIMETIQUES PEPTIDIQUES PARTIELS ET PROCEDES ASSOCIES
Status: Deemed Abandoned and Beyond the Period of Reinstatement - Pending Response to Notice of Disregarded Communication
Bibliographic Data
(51) International Patent Classification (IPC):
  • C07K 14/52 (2006.01)
  • A61K 38/00 (2006.01)
  • A61K 38/04 (2006.01)
  • A61K 38/19 (2006.01)
  • A61P 31/04 (2006.01)
  • A61P 35/00 (2006.01)
  • C07K 7/04 (2006.01)
  • C07K 7/08 (2006.01)
  • C07K 14/00 (2006.01)
  • C07K 19/00 (2006.01)
(72) Inventors :
  • MAYO, KEVIN H. (United States of America)
  • HOYE, THOMAS R. (United States of America)
  • FLADER LAVEY, CAROLEE (United States of America)
(73) Owners :
  • REGENTS OF THE UNIVERSITY OF MINNESOTA
(71) Applicants :
  • REGENTS OF THE UNIVERSITY OF MINNESOTA (United States of America)
(74) Agent: SMART & BIGGAR LP
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2003-02-20
(87) Open to Public Inspection: 2003-08-28
Availability of licence: N/A
Dedicated to the Public: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2003/005106
(87) International Publication Number: WO 2003070751
(85) National Entry: 2004-08-16

(30) Application Priority Data:
Application No. Country/Territory Date
60/359,272 (United States of America) 2002-02-20

Abstracts

English Abstract


Partial peptide mimetics and methods of making and using, wherein the partial
peptide mimetics have a first amino acid sequence comprising ANIKLSVQMKL (SEQ
ID NO: 8), a homolog thereof, or a segment of SEQ ID NO: 8 or a homolog
thereof, a second amino acid sequence comprising IIVKLND (SEQ ID NO:2), a
homolog thereof, or a segment of SEQ ID NO:2 or a homolog thereof, and a
.beta.-turn inducing scaffold bonded between the first and second amino acid
sequences.


French Abstract

L'invention a trait à des mimétiques peptidiques partiels et à des procédés de production et d'utilisation de ceux-ci. Les mimétiques peptidiques partiels contiennent une première séquence d'acides aminés comprenant ANIKLSVQMKL (SEQ ID NO: 8), un homologue de cette dernière, ou un segment de SEQ ID NO: 8 ou un homologue de ce dernier, une seconde séquence d'acides aminés comprenant IIVKLND (SEQ ID NO:2), un homologue de cette dernière, ou un segment de SEQ ID NO:2 ou un homologue de ce dernier, ainsi qu'un squelette inducteur de coude bêta lié entre les première et seconde séquences d'acides aminés.

Claims

Note: Claims are shown in the official language in which they were submitted.


WHAT IS CLAIMED IS:
1. A partial peptide mimetic comprising a first amino acid sequence
comprising: ANIKLSVQMKL (SEQ ID NO:8), a homolog thereof, or a
segment of SEQ ID NO:8 or a homolog thereof; a second amino acid sequence
comprising IIVKLND (SEQ ID NO:2), a homolog thereof, or a segment of SEQ
ID NO:2 or a homolog thereof; and a turn inducing scaffold bonded between
the first and second amino acid sequences; wherein the homologs or segments
provide the partial peptide mimetic with at least one of the following
activities:
inhibition of endothelial cell proliferation in vitro at an IC50 level of less
than
80 µM; inhibition of angiogenesis in vitro at a level of less than 85%
sprouting
in a collagen gel-based assay; or reduction in tumor volume in vivo by at
least
25% relative to a control at the end of an administration period.
2. The partial peptide mimetic of claim 1 wherein the first amino acid
sequence comprises at least three amino acids of SEQ ID NO:8 or a homolog
thereof.
3. The partial peptide mimetic of claim 2 wherein the first amino acid
sequence comprises SEQ ID NO:8, or a homolog thereof, from which a deletion
has been made at the N-terminus by 1, 2, 3, 4, 5, 6, 7, or 8 residues.
4. The partial peptide mimetic of claim 3 wherein the first amino acid
sequence comprises at least one sequence selected from the group consisting of
MKL, QMKL (SEQ ID NO:4), SVQMKL (SEQ ID NO:5), IKLSVQMKL (SEQ
ID NO:6), and NIKLSVQMKL (SEQ ID NO:7).
5. The partial peptide mimetic of claim 1 wherein the first amino acid
sequence comprises ANIKLSVQMKL (SEQ ID NO:8) or a homolog thereof
and the second amino acid sequence comprises IIVKLND (SEQ ID NO:2) or a
homolog thereof.
39

6. The partial peptide mimetic of claim 1 wherein the second amino acid
sequence comprises at least one amino acid of SEQ ID NO:2 or a homolog
thereof.
7. The partial peptide mimetic of claim 6 wherein the second amino acid
sequence comprises SEQ ID NO:2, or a homolog thereof, from which a deletion
has been made at the C-terminus by 1, 2, 3, 4, 5, or 6 residues.
8. The partial peptide mimetic of claim 7 wherein the second amino acid
sequence comprises at least one sequence selected from the group consisting of
I, IIVK (SEQ ID NO:3), and IIVKLN (SEQ ID NO:9).
9. The partial peptide mimetic of claim 8 wherein the first amino acid
sequence comprises at least one sequence selected from the group consisting of
MKL, QMKL (SEQ ID NO:4), SVQMKL (SEQ ID NO:5), IKLSVQMKL (SEQ
ID NO:6), and NIKLSVQMKL (SEQ ID NO:7).
10. The partial peptide mimetic of claim 1 wherein the turn inducing
scaffold is a dibenzofuran-containing scaffold.
11. A partial peptide mimetic comprising a first amino acid sequence
consisting of ANIKLSVQMKL (SEQ ID NO:8) or a segment thereof, a second
amino acid sequence consisting of IIVKLND (SEQ ID NO:2) or a segment
thereof, and a turn inducing scaffold bonded between the first and second
amino acid sequences, wherein the segment of SEQ ID NO:8 is one in which a
deletion has been made at the N-terminus by 1, 2, 3, 4, 5, 6, 7, or 8
residues, and
wherein the segment of SEQ ID NO:2 is one in which a deletion has been made
at the C-terminus by 1, 2, 3, 4, 5, or 6 residues.
12. A partial peptide mimetic comprising a first amino acid sequence having
at least 4 amino acids of the sequence QMKL (SEQ ID NO:4), a second
sequence having at least the amino acid I, and a .beta.-turn inducing scaffold
bonded
40

therebetween, wherein the partial peptide mimetic has at least one of the
following activities: inhibition of endothelial cell proliferation in vitro at
an
IC50 level of less than 80 µM; inhibition of angiogenesis in vitro at a
level of
less than 85% sprouting in a collag-en gel-based assay; or reduction in tumor
volume in vivo by at least 25% relative to a control at the end of an
administration period.
13. The partial peptide mimetic of claim 12 wherein the first amino acid
sequence has at least one of the following:
6 amino acids of the sequence SVQMKL (SEQ ID NO:5);
9 amino acids of the sequence IKLSVQMKL (SEQ ID NO:6);
10 amino acids of the sequence NIKLSVQMKL (SEQ ID NO:7); or
11 amino acids of the sequence ANIKLSVQMKL (SEQ ID NO:8).
14. The partial peptide mimetic of claim 12 wherein the second amino acid
sequence has at least one of the following:
4 amino acids of the sequence IIVK (SEQ ID NO:3);
6 amino acids of the sequence IIVKLN (SEQ ID NO:9); or
9 amino acids of the sequence IIVKLND (SEQ ID NO:2).
15. A partial peptide mimetic selected from the group consisting of:
ANIKLSVQMKL-[DBF]-IIVKLND;
NIKLSVQMKL-[DBF]-IIVKLND;
IKLSVQMKL-[DBF]-IIVKLND;
SVQMKL-[DBF]-IIVKLND;
QMKL-[DBF]-IIVKLND;
MKL-[DBF]-IIVKLND;
L-[DBF]-IIVKLND;
ANIKLSVQMKL-[DBF]-IIVKLN;
ANIKLSVQMKL-[DBF]-IIVK;
ANIKLSVQMKL-[DBF]-I;
SVQMKL-[DBF]-IIVKLN;
41

SVQMKL-[DBF]-IIVK;
SVQMKL-[DBF]-IIV;
SVQMKL-[DBF]-II; and
SVQMKL-(DBF]-I;
wherein DBF is a dibenzofuran .beta.-turn mimetic.
16. A method for inhibiting bacterial infection and/or endotoxemia, the
method comprising contacting cells with an amount of a composition effective
to inhibit the bacterial infection and/or to neutralize endotoxin, wherein the
composition comprises a partial peptide mimetic of claim 1.
17. The method of claim 16 wherein the contacting step occurs in vitro.
18. The method of claim 16 wherein the contacting step occurs in vivo.
19. The method of claim 16 wherein the cells are present in a cell culture, a
tissue, an organ, or an organism.
20. The method of claim 16 wherein the cells are mammalian cells.
21. The method of claim 20 wherein the cells are human cells.
22. The method of claim 16 wherein the partial peptide mimetic neutralizes
endotoxin, is bactericidal, or is both bactericidal and neutralizes endotoxin.
23. A method for decreasing the amount of TNF-.alpha., the method comprising
contacting cells with an amount of a composition effective to decrease the
amount of TNF-.alpha., wherein the composition comprises a partial peptide
mimetic
of claim 1.
24. The method of claim 23 wherein the contacting step occurs in vitro.
42

25. The method of claim 23 wherein the contacting step occurs in vivo.
26. The method of claim 23 wherein the cells are present in a cell culture, a
tissue, an organ, or an organism.
27. The method of claim 23 wherein the cells are mammalian cells.
28. The method of claim 27 wherein the cells are human cells.
29. A method for inhibiting endothelial cell proliferation, the method
comprising contacting cells with an amount of a composition effective to
inhibit
endothelial cell proliferation, wherein the composition comprises a partial
peptide mimetic of claim 1.
30. The method of claim 29 wherein the contacting step occurs in vitro.
31. The method of claim 29 wherein the contacting step occurs in vivo.
32. The method of claim 29 wherein the cells are present in a cell culture, a
tissue, an organ, or an organism.
33. The method of claim 29 wherein the cells are mammalian cells.
34. The method of claim 29 wherein the cells are human cells.
35. A method for inhibiting angiogenic-factor mediated inter-cellular
adhesion molecule expression down-regulation, the method comprising
contacting cells with an amount of a composition effective to inhibit
angiogenic-factor mediated inter-cellular adhesion molecule expression down-
regulation, wherein the composition comprises a partial peptide mimetic of
claim 1.
43

36. The method of claim 35 wherein the contacting step occurs in vitro.
37. The method of claim 35 wherein the contacting step occurs in vivo.
38. The method of claim 35 wherein the cells are present in a cell culture, a
tissue, an organ, or an organism.
39. The method of claim 35 wherein the cells are mammalian cells.
40. The method of claim 35 wherein the cells are human cells.
41. A method for promoting angiogenic-factor mediated inter-cellular
adhesion molecule expression, the method comprising contacting cells with an
amount of a composition effective to promote antiogenic-factor mediated inter-
cellular adhesion molecule expression, wherein the composition comprises a
partial peptide mimetic of claim 1.
42. The method of claim 41 wherein the contacting step occurs in vitro.
43. The method of claim 41 wherein the contacting step occurs in vivo.
44. The method of claim 41 wherein the cells are present in a cell culture, a
tissue, an organ, or an organism.
45. The method of claim 41 wherein the cells are mammalian cells.
46. The method of claim 41 wherein the cells are human cells.
47. A method for inhibiting angiogenesis, the method comprising contacting
cells with an amount of a composition effective to inhibit angiogenesis, the
composition comprising a partial peptide mimetic of claim 1.
44

48. The method of claim 47 wherein the contacting step occurs in vitro.
49. The method of claim 47 wherein the contacting step occurs in vivo.
50. The method of claim 47 wherein the cells are present in a cell culture, a
tissue, an organ, or an organism.
51. The method of claim 47 wherein the cells are mammalian cells.
52. The method of claim 47 wherein the cells are human cells.
53. A method for inhibiting tumorigenesis in a patient, the method
comprising administering to the patient a therapeutically effective amount of
a
composition comprising a partial peptide mimetic of claim 1.
54. A method for inhibiting atherosclerosis in a patient, the method
comprising administering to the patient a therapeutically effective amount of
a
composition comprising a partial peptide mimetic of claim 1.
55. A method for inhibiting restenosis in a patient, the method comprising
administering to the patient a therapeutically effective amount of a
composition
comprising a partial peptide mimetic of claim 1.
56. A method for inhibiting diabetic retinopathy in a patient, the method
comprising administering to the patient a therapeutically effective amount of
a
composition comprising a partial peptide mimetic of claim 1.
57. A method for inhibiting neovascular glaucoma in a patient, the method
comprising administering to the patient a therapeutically effective amount of
a
composition comprising a partial peptide mimetic of claim 1.
45

58. A method for inhibiting rheumatoid arthritis in a patient, the method
comprising administering to the patient a therapeutically effective amount of
a
composition comprising a partial peptide mimetic of claim 1.
59. A method for inhibiting endometriosis in a patient, the method
comprising administering to the patient a therapeutically effective amount of
a
composition comprising a partial peptide mimetic of claim 1.
46

Description

Note: Descriptions are shown in the official language in which they were submitted.


CA 02476427 2004-08-16
WO 03/070751 PCT/US03/05106
Patent
Attorney Docket No. 110.01710201
PARTIAL PEPTIDE MIMETICS AND METHODS
Cross-Reference to Related Applications
The present application claims priority to U.S. Provisional Patent
Application Serial No. 60/359,272, filed on February 20, 2002, which is
incorporated herein by reference.
Background
Angiogenesis, the process of new blood vessel formation, is key to
normal organ development, as well as to various pathological disorders like
cancer, arthritis, diabetic retinopathy and restenosis (A.W. Griffioen et al.,
Biochem. J., 354, 233-242 (2001)). [3pep-25, which has the sequence
ANIKLSVQMKLFKRHLKWKIIVKLNDGRELSLD (SEQ ID NO:1), a
designed cytokine-like ~3-sheet-forming peptide 33mer, is an anti-angiogenic
compound being developed to combat these pathological disorders (K.H. Mayo
et al., Angiogenesis, 4, 45-51 (2001); and S. Liekens et al., J. Biochem.
Pharm.,
61, 253-270 (2001)).
The use of agents that can inhibit angiogenesis, particularly in anti-tumor
research (e.g., M.S. O'Reilly et al., Cell, 88, 277-285 (1997); and T. Boehm
et
al., Nature, 390, 404-407 ( 1997)), has indicated that anti-angiogenic therapy
will be a promising therapeutic modality in the near future. To date, the
search
for angiogenic inhibitors has been focused on controlling two of the processes
that promote angiogenesis: endothelial cell (EC) growth and adhesion (G.
Molema et al., Immunol. Today, l9, 392-394 (1998); and J. Folkman et al.,
Nature Med., 1, 27-31 (1995). Targeting EC as an anti-tumor treatment is
attractive primarily because EC are more accessible than are other cells to
pharmacologic agents delivered via the blood, and EC are genetically stable
and
are not easily mutated into drug resistant variants.
1

CA 02476427 2004-08-16
WO 03/070751 PCT/US03/05106
Most anti-angiogenic agents have been discovered by identifying
endogenous molecules, primarily proteins, which inhibit EC growth. This
traditional approach has produced a number of anti-angiogenics, such as
platelet
factor-4 (PF4), thrombospondin, tumor necrosis factor-a (TNF-a, depending on
S its concentration), interferon-y inducible protein-10, angiostatin,
endostatin and
vasostatin and bactericidal-permeability increasing (BPI) protein. See, for
example, M.S. O'Reilly et al., Cell, 88, 277-285 (1997); S.K. Gupta et al., J.
Cell Biol.,127, 1121-1127 (1994); S.S. Tolsma et al., J. Cell Biol., 122, 497-
511 (1993); N. Sato et al., Japan Natl. Cancer Inst., 76, 1113- 1121 (1986);
V.J.
Palombella et al., J. Biol. Chem., 264, 18128-18136 (1989); B. Robaye et al.,
Am. J. Pathol.,138, 447-453 (1991); A.D. Luster et al., J. Exp. Med.,182, 219-
231 (1995); M.S. O'Reilly et al., Cell, 79, 315-328 (1994); S.E. Pike et al.,
J.
Exp. Med.,188, 2349-2356 (1998); and D.W.J. Van der Schaft et al., Blood, 96,
176-181 (2000). About forty anti-angiogenic agents are currently known.
Most anti-angiogenic proteins are compositionally similar, having a
relatively high incidence of hydrophobic and positively charged residues and
are
folded primarily as (3-sheets (A.R. Mire Sluis et al., J. Immunol. Methods,
200,
1-16 (1997)): interleukin-1 (IL-1), tumor necrosis factor (TNF), lymphotoxin
(LT or TNF-(3), transforming growth factor-(3 (TGF-(3), endostatin. See, for
example, J.P. Priestle et al., Proc. Natl. Acad. Sci. USA, 86, 9667-9671
(1989);
E.Y. Jones et al., Nature, 338, 225-228 (1989); M.J. Eck et al., J. Biol.
Chem.,
267, 2119- 2122 (1992); S. Daopin et al., Science, 257, 369-373 (1992); and E.
Hohenester et al., EMBOJ.,17, 1656-1664 (1998).
Recently, a designed amphipathic ~3-sheet-forming peptide 33mer, ~3pep-
25 was shown to be a potent inhibitor of EC growth and angiogenesis. ~3pep-25
is more effective at inhibiting EC growth than PF4 and several other well-
known angiogenesis inhibitors such as angiostatin, endostatin, AGM-1470 and
thrombospondin-1. (3pep-25 is believed to act by specifically blocking
adhesion
and migration of angiogenically-activated EC, leading to apoptosis and
ultimately to inhibition of angiogenesis in vitro and in vivo and inhibits
tumor
growth by up to 80% in various models. See, for example, D.W.J. Van der
2

CA 02476427 2004-08-16
WO 03/070751 PCT/US03/05106
Schaft et al., Faseb J.,16, 1991-1993 (2002); and R.P. Dings et al., Cancer
Res., 63, 382-385 (2003).
For the smart design of smaller compounds, the identification of specific
amino acid residues and their spatial relationships are used. One of the main
goals among structural biologists and pharmaceutical chemists is to develop
small molecules and potentially more effective anti-tumor agents.
Nevertheless,
for anti-angiogenic proteins such structure-activity relationships (SAR),
i.e.,
specific residues and conformations which impart activity, are sorely needed,
and even the analysis of high-resolution molecular structures of a number of
anti-angiogenic proteins, e.g., endostatin, PF4, and BPI, has yet to provide
this
information. See, for example, E. Hohenester et al., EMBD J., 17, 1656-1664
(1998); K.H. Mayo et al., Biochemistry, 34, 11399-11409 (1995); and L.J.
Beamer et al., Science, 276, 1861-1864 (1997).
1 S Summary
From the structures of angiogenesis inhibitors that are known it appears
that most anti-angiogenic proteins are compositionally similar. They show a
relatively high incidence of hydrophobic and positively charged residues and a
(3-sheet. (3pep-25 also falls into this category as an amphipathic (3-sheet-
forming
peptide 33-mer. It is believed that this (3-sheet conformation is necessary
for the
bioactivity of the 33-mer (3pep-25. The present invention is directed to the
identification of specific residues and conformations that are responsible for
activity, leading to the design of partial peptide mimetics of (3pep-25.
As used herein, a peptide mimetic is a compound that mimics a peptide.
The use of the adjective "partial" indicates that it is not a full mimetic
because it
contains some peptide segments, i.e., amino acids in a sequence.
The present invention relates to partial peptide mimetics and methods
for using such compounds that include segments (i.e., portions) of the (ipep-
25
sequence, which is ANIKLSVQMKLFKRHLKWKIIVKLNDGRELSLD (SEQ
ID NO:1 ), or homologs thereof.
In particular, the partial peptide mimetics of the present invention are
designed to include all or segments of the sequences ANIKLSVQMKL (SEQ ID
3

CA 02476427 2004-08-16
WO 03/070751 PCT/US03/05106
N0:8) or a homolog thereof, and IIVKLND (SEQ ID N0:2) or a homolog
thereof, with a ~3-turn inducing scaffold bonded therebetween (e.g.,
covalently
bonded). Segments of these sequences (SEQ ID N0:8 and SEQ ID N0:2 or
homologs thereof) include one or more amino acids. Segments of SEQ ID
N0:8 and SEQ ID N0:2 as well as homologs of the segments and the full
sequences preferably provide the partial peptide mimetic with at least one of
the
following activities: inhibition of endothelial cell proliferation in vitro at
an
IC50 level of less than 80 ~,M; inhibition of angiogenesis in vitro at a level
of
less than 85% sprouting in a collagen gel-based assay; or reduction in tumor
volume in vivo (e.g., in an animal model) by at least 25% relative to a
control at
the end of an administration period.
In certain embodiments, segments of SEQ ID N0:8 or homologs thereof
preferably include at least three amino acids of SEQ ID N0:8 or homologs
thereof (preferably, contiguous amino acids), and more preferably include at
least six amino acids of SEQ ID N0:8 or homologs thereof (preferably,
contiguous amino acids). In certain embodiments, segments of SEQ ID N0:8 or
homologs thereof include those in which deletions have been made at the N-
terminus preferably by 1, 2, 3, 4, 5, 6, 7, or 8 residues, more preferably by
1, 2,
3, 4, or 5 residues. Examples of segments of SEQ ID N0:8 that can be used in
forming the partial peptide mimetics of the present invention include MKL,,
QMKL (SEQ ID N0:4), SVQMKL (SEQ ID NO:S), IKLSVQMKL (SEQ ID
N0:6), and NIKLSVQMKL (SEQ ID N0:7). Particularly preferred such
segments include, for example, SVQMKL (SEQ ID NO:S), IKLSVQMKL
(SEQ ID N0:6), and NIKLSVQMKL (SEQ ID N0:7).
In certain embodiments, segments of SEQ ID N0:2 or homologs thereof
include at least one amino acid of SEQ ID N0:2 or homologs thereof, and
preferably at least four amino acids of SEQ ID N0:2 or homologs thereof
(preferably, contiguous amino acids). In certain preferred embodiments,
segments of SEQ ID N0:2 or homologs thereof include those in which deletions
have been made at the C-terminus by 1, 2, 3, 4, 5, or 6 residues. Particularly
preferred segments of SEQ ID N0:2 that can be used in forming the partial
4

CA 02476427 2004-08-16
WO 03/070751 PCT/US03/05106
peptide mimetics of the present invention include I, IIVK (SEQ ID N0:3), and
IIVKLN (SEQ ID N0:9).
The turn inducing scaffold can be a chemical scaffold, such as a
dibenzofuran-containing scaffold.
Particularly preferred partial peptide mimetics of the present invention
include at least the 7 amino acids IIVKLND (SEQ ID N0:2) in combination
with at least the 4 amino acids QMKL (SEQ ID N0:4) with a turn inducing
scaffold, such as a chemical scaffold like a dibenzofuran-containing scaffold,
bonded therebetween. Other preferred partial peptide mimetics include at least
SEQ ID N0:2 and at least the 6 amino acids SVQMKL (SEQ ID N0:5) with a
/turn inducing scaffold bonded therebetween. Other preferred partial peptide
mimetics include at least SEQ ID N0:2 and at least the 9 amino acids
IKLSVQMKL (SEQ ID N0:6) with a,l~turn inducing scaffold bonded
therebetween. Other preferred partial peptide mimetics include at least SEQ ID
N0:2 and at least the 10 amino acids NILKSVQMKL (SEQ ID N0:7) with a ~
turn inducing scaffold bonded therebetween. Other preferred partial peptide
mimetics include at least SEQ ID N0:2 and at least the 11 amino acids
ANILKSVQMKL (SEQ ID N0:8) with a,(~turn inducing scaffold bonded
therebetween.
Other preferred partial peptide mimetics include at least
ANIKLSVQMKL (SEQ ID N0:8) and at least the amino acid I with a /turn
inducing scaffold bonded therebetween. Other preferred partial peptide
mimetics include at least SEQ ID N0:8 and at least the 4 amino acids IIVK
(SEQ ID N0:3) with a turn inducing scaffold bonded therebetween. Other
preferred partial peptide mimetics include at least SEQ ID N0:8 and at least
the
6 amino acids IIVKLN (SEQ ID N0:9) with a turn inducing scaffold bonded
therebetween.
Other preferred partial peptide mimetics include at least SVQMKL
(SEQ ID N0:5) and at least the amino acid I with a turn inducing scaffold
therebetween. Other preferred partial peptide mimetics include at least SEQ ID
5

CA 02476427 2004-08-16
WO 03/070751 PCT/US03/05106
NO:S and at least the 6 amino acids IIVKLN (SEQ ID N0:9) with a turn
inducing scaffold bonded therebetween.
This invention also relates to methods of using at least one of the partial
peptide mimetics disclosed herein. Preferably, the methods are for treatment
of
various conditions in vivo, although in vitro methods are also desirable.
Typically, such methods involve the use of a composition that includes at
least
one of the partial peptide mimetics and optionally a carrier, preferably a
pharmaceutically acceptible carrier.
Such methods include, for example, treating a bacterial infection or
endotoxic shock. In one embodiment, the partial peptide mimetic neutralizes
endotoxin, in another the partial peptide mimetic is bactericidal, and in
another
the partial peptide mimetic is both bactericidal and neutralizes endotoxin.
Such
partial peptide mimetics can also be used to inhibit bacterial infection or
endotoxic shock in a cell culture.
This invention also relates to a method for inhibiting TNF-a levels,
inhibiting endothelial cell proliferation, promoting inter-cellular adhesion
molecule (ICAM) expression, inhibiting inter-cellular adhesion molecule
(ICAM) expression down regulation, inhibiting tumorigenesis, and inhibiting
angiogenesis. Such methods can be carried out in vivo (e.g., for treating a
mammal) or in vitro (e.g., in a cell culture). Similarly, such partial peptide
mimetics can be used to inhibit pathologic disorders such as atherosclerosis,
restenosis, diabetic retinopathy, neovascular glaucoma, rheumatoid arthritis,
and
endometriosis.
As used herein "a " "an " "the " "at least one " and "one or more" are
> > > > >
used interchangeably.
"Amino acid" is used herein to refer to a chemical compound with the
general formula: NH2---CRH---COON, where R, the side chain, is H or an
organic group. Where R is organic, R can vary and is either polar or nonpolar
(i.e., hydrophobic). The amino acids of this invention can be naturally
occurring
or synthetic (often referred to as nonproteinogenic). As used herein, an
organic
group is a hydrocarbon group that is classified as an aliphatic group, a
cyclic
group or combination of aliphatic and cyclic groups. The term "aliphatic
group"
6

CA 02476427 2004-08-16
WO 03/070751 PCT/US03/05106
means a saturated or unsaturated linear or branched hydrocarbon group. This
term is used to encompass alkyl, alkenyl, and alkynyl groups, for example. The
term "cyclic group" means a closed ring hydrocarbon group that is classified
as
an alicyclic group, aromatic group, or heterocyclic group. The term "alicyclic
group" means a cyclic hydrocarbon group having properties resembling those of
aliphatic groups. The term "aromatic group" refers to mono- or polycyclic
aromatic hydrocarbon groups. As used herein, an organic group can be
substituted or unsubstituted. One letter and three letter symbols are used
herein
to designate the naturally occurring amino acids. Such designations including
R
or Arg, for Arginine, K or Lys, for Lysine, G or Gly, for Glycine, and X for
an
undetermined amino acid, and the like, are well known to those skilled in the
art.
The terms "polypeptide" and "peptide" as used herein are used
interchangeably and refer to a compound that includes two or more amino acids.
1 S These terms do not connote a specific length of amino acids (e.g., herein,
an
"amino acid sequence" can include just one amino acid).
The term "partial peptide mimetic" is used herein to refer to a compound
that mimics a peptide, and contains some peptide segments, i.e., amino acids
in
a sequence. At times these compounds are simply referred to as "peptides" or
"polypeptides."
7

CA 02476427 2004-08-16
WO 03/070751 PCT/US03/05106
The following abbreviations are used throughout the application:
A = Ala = Alanine ~ T = Thr = Threonine
V = Val = Valine ~ C = Cys = Cysteine
L = Leu = Leucine ~ Y = Tyr = Tyrosine
I = Ile = Isoleucine ~ N = Asn = Asparagine
P = Pro = Proline ~ Q = Gln = Glutamine
F = Phe = Phenylalanine ~ D = Asp = Aspartic Acid
W = Trp = Tryptophan ~ E = Glu = Glutamic Acid
M = Met = Methionine ~ K = Lys = Lysine
G = Gly = Glycine ~ R = Arg = Arginine
S = Ser = Serine ~ H = His = Histidine
Other abbreviations used herein include: DBF, dibenzofuran; EC,
endothelial cell; EAM, endothelial adhesion molecule; HUVEC, human
umbilical vein EC; PBS, phosphate buffered saline; HPLC, high performance
liquid chromatography; bFGF, basic fibroblast growth factor; VEGF, vascular
endothelial growth factor; PF4, platelet factor-4; BPI, bactericidal-
permeability
increasing protein; NMR, nuclear magnetic resonance; GC-LRMS, gas
chromatography-liquid chromatography mass spectrometry; IR, infrared
spectroscopy; SPPS, solid phase peptide synthesis; BOP/HOBT, benzotriazol-1-
yloxy)tris(dimethylamino) phosphonium hexafluorophosphate/1-
hydroxybenzotriazole; DPC, dodecylphosphocholine.
Brief Description of the Drawings
Figure 1 (A). Sequences for the dodecapeptide walk-throughs below the
amino acid sequence of (3pep-25. Proliferation of bFGF-stimulated (10 ng/ml)
human umbilical vascular EC (HUVEC) cultures was measured by
quantification of 3H-thymidine incorporation. Proliferation is expressed as
mean
8

CA 02476427 2004-08-16
WO 03/070751 PCT/US03/05106
counts per minute (cpm) of quadruplicate cultures in three independent
experiments (~ SEM). EC proliferation results from alanine scanning (tested at
25 ~,M dose) are expressed in bar graph format as the percentage of
proliferating
EC (the arithmetic mean counts per minute (cpm) of triplicate cultures)
relative
to control cultures. For walk-through peptides, results are given as ICso
values
from dose response curves; only ICSO values for dodecapeptides with
significant
activity relative to ~3pep-25 are given.
Figure 1 (B). Overall fold for ~ipep-25 (3 ~i-strands with 2 turns), shows
with functionally key residues boxed-in.
Figure 1 (C) shows the parent DBF analog ( 11 DBF7 or CF-8) with the
introduction of the scaffold.
Figure 2. The superposition of 6DBF7 derived from NMR analysis is
shown in Figure 2(A), with the chemical 3D structure illustrated in Figure
2(B).
Figure 3. Anti-proliferative effects of DBF analogs on bFGF-activated
1 S EC are plotted as dose response curves.
Figure 4. MA148 tumor bearing mice were treated with the optimal
dose of (3pep-25 (10 mg/kg/day) or equivalent doses of 11DBF7 or 6DBF7. In
Figure 4(A) treatment was initiated at the time of inoculation with MA148
cells.
In Figure 4(B) an intervention study is shown where tumors were allowed to
establish to a palpable size before treatment was initiated. In either study,
control groups of animals were treated with PBS containing human serum
albumin to control for protein content. Tumor volumes (for all groups n = 11,
t
SEM) are plotted as mm3 vs. days post inoculation. The inserts show the body
weight development of the mice during the study as a measurement of overall
toxicity.
Detailed Description of Illustrative Embodiments of the Invention
The present invention provides partial peptide mimetics and methods of
using such partial peptide mimetics. These partial peptide mimetics include
segments (i.e., portions) of the (3pep-25 sequence, which is
ANIKLSVQMKLFKRHLKWKIIVKLNDGRELSLD (SEQ ID NO:l), or
9

CA 02476427 2004-08-16
WO 03/070751 PCT/US03/05106
homologs thereof. These partial peptide mimetics preferably include at least 7
amino acid residues. Although there is no upper limit to the number of amino
acid residues, it is desirable for the partial peptide mimetics to be small
and
approach the size of conventional therapeutic small organic molecules.
In particular, the partial peptide mimetics of the present invention are
designed to include all or segments of the sequences ANIKLSVQMKL (SEQ ID
N0:8) or a homolog thereof, and IIVKLND (SEQ ID N0:2) or a homolog
thereof, with a /.turn inducing scaffold bonded therebetween (e.g., covalently
bonded). Segments of these sequences (SEQ ID N0:8 and SEQ ID N0:2 or
homologs thereof) include one or more amino acids.
Segments of SEQ ID N0:8 and SEQ ID N0:2 as well as homologs of
the segments and the full sequences preferably provide the partial peptide
mimetic with at least one of the following activities: inhibition of
endothelial
cell proliferation in vitro at an ICSO level of less than 80 ~,M (more
preferably
less than 50 ~,M, and even more preferably less than 25 ~M) as determined
using the assay described in the Examples Section; inhibition of angiogenesis
in
vitro at a level of less than 85% sprouting (more preferably less than 75%
sprouting, even more preferably 50% sprouting, and even more preferably less
than 35% sprouting) as determined using the collagen gel-based assay described
in the Examples Section; or reduction in tumor volume in vivo by at least 25%
(more preferably by at least 50%, and even more preferably by at least 70%)
relative to a control at the end of an administration period as determined
using
the assay with an animal model described in the Examples Section. These
values can be determined by one of skill in the art and are typically average
values.
In certain embodiments, segments of SEQ ID N0:8 or homologs thereof
preferably include at least three amino acids of SEQ ID N0:8 or homologs
thereof (preferably, contiguous amino acids), and more preferably include at
least six amino acids of SEQ ID N0:8 or homologs thereof (preferably,
contiguous amino acids). In certain embodiments, segments of SEQ ID N0:8 or
homologs thereof include those in which deletions have been made at the N-
terminus preferably by 1, 2, 3, 4, 5, 6, 7, or 8 residues, more preferably by
1, 2,

CA 02476427 2004-08-16
WO 03/070751 PCT/US03/05106
3, 4, or 5 residues, and even more preferably by 5 residues. Examples of
segments of SEQ ID N0:8 that can be used in forming the partial peptide
mimetics of the present invention include MKL, QMKL (SEQ ID N0:4),
SVQMKL (SEQ ID NO:S), IKLSVQMKL (SEQ ID N0:6), and
NIKLSVQMKL (SEQ ID N0:7). Particularly preferred such segments include,
for example, SVQMKL (SEQ ID NO:S), IKLSVQMKL (SEQ ID N0:6), and
NIKLSVQMKL (SEQ ID N0:7).
In certain embodiments, segments of SEQ ID N0:2 or homologs thereof
include at least one amino acid of SEQ ID N0:2 or homologs thereof, and
preferably at least four amino acids of SEQ ID N0:2 or homologs thereof
(preferably, contiguous amino acids). In certain preferred embodiments,
segments of SEQ ID N0:2 include those in which deletions have been made at
the C-terminus by l, 2, 3, 4, 5, or 6 residues. Particularly preferred
segments of
SEQ ID N0:2 that can be used in forming the partial peptide mimetics of the
present invention include I, IIVK (SEQ ID N0:3), and IIVKLN (SEQ ID N0:9).
The /.turn inducing scaffold can be a chemical scaffold, such as a
dibenzofuran-containing scaffold or biphenyl- containing scaffold. Preferably,
it is a dibenzofuran-containing scaffold. More preferably, the dibenzofuran-
containing scaffold is that shown in the Examples Section (Scheme 1) and is
referred to herein as "DBF." It should be understood that the length of the
hydrocarbon chains off the phenyl rings in the scaffold and attached to the
peptides can vary in length.
Such partial peptide mimetics are active with respect to a number of
biological activities. This is exemplified by the data shown herein with
respect
to specific partial peptide mimetics composed of a (3-sheet-inducing
biphenylfuran scaffold and two short amino acid sequences from ~ipep-25. They
can be as effective as (3pep-25 at inhibiting endothelial cell proliferation
and
angiogenesis in vitro. For example, in a mouse xenograft model for ovarian
carcinoma, two variations of the partial mimetic are observed to be slightly
more effective than (3pep-25 by reducing tumor volume by up to 85%.
Immunohistochemical staining indicates that antitumor activity is mediated by
significant reduction in vessel density and endothelial cell proliferation, as
well
11

CA 02476427 2004-08-16
WO 03/070751 PCT/US03/05106
as increased cell apoptosis. Overall, the partial peptide mimetic demonstrates
improved bioavailability over ~3pep-25 and the potential for obtaining an
orally
active, small molecule of an angiogenesis inhibitory protein. Because of this
anti-angiogenic behavior it is believed that such partial peptide mimetics can
be
used to inhibit pathologic disorders such as atherosclerosis, restenosis,
diabetic
retinopathy, neovascular glaucoma, rheumatoid arthritis, and endometriosis.
This can be evaluated readily by one of skill in the art using appropriate in
vitro
and in vivo models.
Particularly preferred partial peptide mimetics include 6DBF1, 6DBF7,
and 11DBF7, of which even more preferred are 6DBF7 and 11DBF7. The
sequences of these DBF analogs (which are partial peptide mimetics) are shown
in Table 1.
Such partial peptide mimetics can be in their free acid form or they can
be amidated at the C-terminal carboxylate group. The present invention also
includes homologs of the peptide sequences listed herein, which typically have
structural similarity with such peptides. A "homolog" of a polypeptide
includes
one or more conservative amino acid substitutions, which are selected from
other members of the class to which the amino acid belongs. For example, it is
well-known in the art of protein biochemistry that an amino acid belonging to
a
grouping of amino acids having a particular size or characteristic (such as
charge, hydrophobicity and hydrophilicity) can generally be substituted for
another amino acid without substantially altering the structure of a
polypeptide.
For the purposes of this invention, conservative amino acid substitutions
are defined to result from exchange of amino acids residues from within one of
the following classes of residues: Class I: Ala, Gly, Ser, Thr, and Pro
(representing small aliphatic side chains and hydroxyl group side chains);
Class
II: Cys, Ser, Thr, and Tyr (representing side chains including an -OH or -SH
group); Class III: Glu, Asp, Asn, and Gln (carboxyl group containing side
chains): Class IV: His, Arg, and Lys (representing basic side chains); and
Class
V: Ile, Val, Leu, Phe, Met, Phe, Trp, Tyr, and His (representing hydrophobic
side chains). The classes also include related amino acids such as 3Hyp and
4Hyp in Class I; homocysteine in Class II; 2-aminoadipic acid, 2-aminopimelic
12

CA 02476427 2004-08-16
WO 03/070751 PCT/US03/05106
acid, y carboxyglutamic acid, (3-carboxyaspartic acid, and the corresponding
amino acid amides in Class III; ornithine, homoarginine, N-methyl lysine,
dimethyl lysine, trimethyl lysine, 2,3-diaminopropionic acid, 2,4-
diaminobutyric
acid, homoarginine, sarcosine and hydroxylysine in Class IV; substituted
phenylalanines, norleucine, norvaline, 2-aminooctanoic acid, 2-aminoheptanoic
acid, statine and ~3-valine in Class V; and naphthylalanines, substituted
phenylalanines, tetrahydroisoquinoline-3-carboxylic acid, and halogenated
tyrosines in Class V.
Polypeptide homologs, as that term is used herein, also include modified
I O polypeptides. Modifications of polypeptides of the invention include
chemical
and/or enzymatic derivatizations at one or more constituent amino acid,
including side chain modifications, backbone modifications, and N- and C-
terminal modifications including acetylation, hydroxylation, methylation,
amidation, and the attachment of carbohydrate or lipid moieties, cofactors,
and
the like.
Such homologs provide partial peptide mimetics with one or more of the
biological activities described herein.
The partial peptide mimetics, particularly the peptide portions of the
partial peptide mimetics of the invention, may be synthesized by the solid
phase
method using standard methods based on either t-butyloxycarbonyl (BOC) or 9-
fluorenylmethoxy-carbonyl (FMOC) protecting groups. This methodology is
described by G.B. Fields et al. in Synthetic Peptides: A User's Guide, W.M.
Freeman & Company, New York, NY, pp. 77-183 (1992).
A preferred partial peptide mimetic is characterized by having at least
one of the biological activities described herein. The biological activity of
a
polypeptide can be determined, for example, as described herein or by methods
well known to one of skill in the art.
Compositions comprising one or more of the partial peptide mimetics of
this invention with an optional carrier (e.g., a pharmaceutically acceptable
carrier) can be added to cells in culture or used to treat patients, such as
mammals. Where the partial peptide mimetics are used to treat a patient, the
partial peptide mimetic is preferably combined in a pharmaceutical composition
13

CA 02476427 2004-08-16
WO 03/070751 PCT/US03/05106
with a pharmaceutically acceptible carrier, such as a larger molecule to
promote
stability or a pharmaceutically acceptible buffer that serves as a carrier.
Treatment can be prophylactic or therapeutic. Thus, treatment can be
initiated before, during, or after the development of the condition (e.g.,
bacterial
infection or endotoxemia). As such, the phrases "inhibition of or "effective
to
inhibit" a condition such as bacterial infection and/or endotoxemia, for
example,
includes both prophylactic and therapeutic treatment (i.e., prevention and/or
reversal of the condition).
The partial peptide mimetics of the present invention can be
administered alone or in a pharmaceutically acceptable buffer, as an antigen
in
association with another protein, such as an immunostimulatory protein or with
a protein carrier such as, but not limited to, keyhole limpet hemocyanin
(KLH),
bovine serum albumin (BSA), ovalbumin, or the like.
Partial peptide mimetics can be conjugated to other proteins using
standard methods such as activation of the Garner molecule with a
heterobifunctional sulfosuccinimidyl 4-(n-maleimidomethyl) cyclohexane-1-
carboxylate reagent. Cross-linking of an activated carrier to a peptide can
occur
by reaction of the maleimide group of the carrier with the sulfhydryl group of
a
peptide containing a cysteine residue. Conjugates can be separated from
nonconjugated molecules through the use of gel filtration column
chromatography or other methods known in the art.
The partial peptide mimetics can be combined with a variety of
physiological acceptable carriers for delivery to a patient including a
variety of
diluents or excipients known to those of ordinary skill in the art. For
example,
for parenteral administration, isotonic saline is preferred. For topical
administration a cream, including a carrier such as dimethylsulfoxide (DMSO),
or other agents typically found in topical creams that do not block or inhibit
activity of the peptide, can be used. Other suitable carriers include, but are
not
limited to alcohol, phosphate buffered saline, and other balanced salt
solutions.
The partial peptide mimetics of this invention that demonstrate
biological activity can be administered in a variety of ways, including
intravenously, topically, orally, and intramuscularly to a variety of mammals,
14

CA 02476427 2004-08-16
WO 03/070751 PCT/US03/05106
including humans, mice and rabbits. The partial peptide mimetics can be
administered as a single dose or in multiple doses. Preferably the dose is an
effective amount as determine by the standard methods described herein and
includes about 1 microgram to about l, 000 micrograms pretreatment, more
preferably about 50 to about 250 micrograms pretreatment. Those skilled in the
art of clinical trials will be able to optimize dosages of particular partial
peptide
mimetics through standard trial studies.
Preferred partial peptide mimetics are anti-angiogenic. Angiogenesis is
crucial to numerous biological functions in the body, from normal processes
like
embryogenesis and wound healing to abnormal processes like tumor growth,
arthritis, restenosis, atherosclerosis, diabetic retinopathy, neovascular
glaucoma,
and endometriosis. The use of agents that can inhibit angiogenesis in vitro
and
in vivo, particularly in anti-tumor research, has indicated that anti-
angiogenic
therapy will be a promising therapeutic modality in the future. The search for
angiogenic inhibitors has been focused on controlling two of the processes
that
promote angiogenesis: endothelial cell (EC) growth and adhesion primarily
because ECs are more accessible than are other cells to pharmacologic agents
delivered via the blood and ECs are genetically stable and are not easily
mutated
into drug resistant variants. Most anti-angiogenic agents have been discovered
by identifying endogenous molecules, primarily proteins, which inhibit EC
growth. This traditional approach has produced a number of anti-angiogenics,
such as platelet factor-4 (PF4), thrombospondin, tumor necrosis factor (TNF),
interferon-'y inducible protein-10, angiostatin, endostatin, vasostatin, and
bactericidal-permeability increasing (BPI) protein. In total, about forty anti-
angiogenic agents, identified using various approaches, are currently known.
It has also been postulated that tumor growth can be controlled by
deprivation of vascularization (Folkman J. Natl. Cancer. Inst., 82, 4-6
(1990);
Folkman et al., J. Biol. Chem., 267, 10931-10934 (1992)). A growing number
of endogenous inhibitors of angiogenesis such as platelet factor-4 (PF4),
interferon-yinducible protein-10 (IP-10), thrombospondin-1 (TSP-1),
angiostatin, as well as synthetic agents, e.g., thalidomide, TNP-470, and
metalloproteinase inhibitors have been described. Some of these agents are

CA 02476427 2004-08-16
WO 03/070751 PCT/US03/05106
currently being tested in phase I/II clinical trials. Previous research
described in
Griffioen et al., Blood, 88, 667-673 (1996), and Griffioen et al., Cancer
Res.,
56, 1111-1117 ( 1996) has shown that pro-angiogenic factors in tumors induce
down-regulation of adhesion molecules on endothelial cells in the tumor
vasculature and induce anergy to inflammatory signals such as tumor necrosis
factor a (TNF-a), interleukin-1, and interferon-y. EC exposed to vascular
endothelial cell growth factor (VEGF) (Griffioen et al., Blood, 88, 667-673
(1996)) and basic fibroblast growth factor (bFGF) (Griffioen et al., Blood,
88,
667-673 (1996); and Melder et al., Nature Med., 2, 992-997 (1996)) have a
severely hampered up-regulation of intercellular adhesion molecule-1 (ICAM-1)
and induction of vascular cell adhesion molecule-1 (VCAM-1 ) and E-selectin.
This phenomenon, which was named tumor-induced EC anergy, is one way in
which tumors with an angiogenic phenotype may escape infiltration by cytotoxic
leukocytes.
Because angiogenesis-mediated down-regulation of endothelial
adhesion molecules (EAM) may promote tumor outgrowh by avoiding the
immune response (Griffioen et al., Blood, 88, 667-673 (1996); Kitayama et al.,
Cancer. Res., 54 4729-4733 (1994); and Piali et al., J. Exp. Med ,181, 811-816
(1995)), it is believed that inhibition of angiogenesis would overcome the
down-
regulation of adhesion molecules and the unresponsiveness to inflammatory
signals. In support of this hypothesis, a relation between E-selectin up-
regulation and the angiostatic agent AGM-1470 has been reported (Budson et
al., Biochem. Biophys. Res. Comm., 225, 141-145 (1996)). It has also been
shown that inhibition of angiogenesis by PF-4 up-regulates ICAM-1 on bFGF-
simulated EC. In addition, inhibition of angiogenesis by PF4 overcomes the
angiogenesis-associated EC anergy to inflammatory signals.
Thus, the present invention provides a method for inhibiting endothelial
cell proliferation in a patient (e.g., a mammal such as a human). This
involves
administering to a patient an amount of a composition (typically a
pharmaceutical composition) effective to inhibit the growth or endothelial
cells,
wherein the composition includes one or more partial peptide mimetics
described herein. Analogously, the present invention provides a method for
16

CA 02476427 2004-08-16
WO 03/070751 PCT/US03/05106
inhibiting endothelial cell proliferation in vitro (e.g., in a cell culture).
This
method involves contacting cells with an amount of a composition effective to
prevent and/or reduce the growth of endothelial cells, wherein the composition
includes one or more partial peptide mimetics described herein.
For determining the amount of endothelial cell proliferation in vivo,
various methods known to one of skill in the art could be used. For example,
for evaluation of endothelial cell growth in tumors, tissue sections can be
appropriately stained to quantify vessel density. For determining the amount
of
endothelial cell proliferation in vitro, an EC Proliferation Assay can be used
that
involves the uptake of tritiated thymidine by cells in cell culture. A partial
peptide mimetic that is "active" for inhibiting endothelial cell proliferation
is
preferably one that causes an at least 10% reduction in endothelial cell
proliferation at a concentration lower than 10~ M. Alternatively, inhibition
of
endothelial cell proliferation for an "active" partial peptide mimetic in
vitro is
preferably at an IC50 level of less than 80 ~.M (more preferably less than 50
~,M, and even more preferably less than 25 ~M) as determined using the assay
described in the Examples Section.
The present invention also provides a method for inhibiting angiogenic-
factor mediated inter-cellular adhesion molecule (ICAM) expression down-
regulation (and/or promoting ICAM expression) in a patient (e.g., a mammal
such as a human). This involves administering to a patient an amount of a
composition effective to prevent and/or reduce the amount of ICAM expression
down-regulation, wherein the composition includes one or more partial peptide
mimetics described herein. Analogously, the present invention provides a
method for inhibiting angiogenic-factor mediated inter-cellular adhesion
molecule expression down-regulation (and/or promoting ICAM expression) in
vitro (e.g., in a cell culture). This method involves contacting cells with an
amount of a composition effective to prevent and/or reduce the amount of
ICAM expression down-regulation, wherein the composition includes one or
more partial peptide mimetics described herein.
The present invention provides a method for inhibiting angiogenesis
(i.e., new blood vessel formation) in a patient (e.g., a mammal such as a
17

CA 02476427 2004-08-16
WO 03/070751 PCT/US03/05106
human). This involves administering to a patient an amount of a composition
effective to prevent and/or reduce angiogenesis, wherein the composition
includes one or more partial peptide mimetics described herein. Analogously,
the present invention provides a method for inhibiting angiogenesis in vitro
(e.g., in a cell culture). This method involves contacting cells with an
amount of
a composition effective to prevent and/or reduce angiogenesis, wherein the
composition includes one or more partial peptide mimetics described herein.
For determining the amount of angiogenesis in vivo, various methods
known to one of skill in the art could be used. For example, for evaluation of
angiogenesis in tumors, tissue sections can be appropriately stained to
quantify
vessel density. For determining the amount of angiogenesis in vitro, an
Angiogenesis Assay can be used that involves the disappearance of EC
sprouting in cell culture. A polypeptide that is "active" for angiogenesis
inhibition is preferably one that causes an at least 10% reduction in
endothelial
cell sprouting at a concentration lower than 10~ M. Alternatively, inhibition
of
angiogenesis for a partial peptide mimetic in vitro is preferably at a level
of less
than 85% sprouting (more preferably less than 75% sprouting, even more
preferably 50% sprouting, and even more preferably less than 35%) as
determined using the collagen gel-based assay described in the Examples
Section.
Similarly, such anti-angiogenic compositions can be used to control
pathologic disorders such as atherosclerosis, restenosis, diabetic
retinopathy,
neovascular glaucoma, rheumatoid arthritis, and endometriosis. This can be
demonstrated using standard techniques and models known to one of skill in the
art.
The present invention provides a method for inhibiting tumorigenesis in
a patient (e.g., a mammal such as a human). This involves administering to a
patient an amount of a composition effective to prevent and/or reduce tumor
growth, wherein the composition includes one or more partial peptide mimetics
described herein. Methods of determining the inhibition of tumorigenesis are
well known to those of skill in the art, including evaluation of tumor
shrinkage,
survival, etc.
18

CA 02476427 2004-08-16
WO 03/070751 PCT/US03/05106
The present invention provides a method for treating bacterial infection
and/or endotoxemia in a patient (e.g., a mammal such as a human). This
involves administering to a patient an amount of a composition effective to
inhibit the bacterial infection and/or to neutralize endotoxin, wherein the
pharmaceutical composition includes one or more partial peptide mimetics
described herein. Analogously, the present invention provides a method for
inhibiting bacterial infection and/or endotoxemia in vitro (e.g., in a cell
culture).
This method involves contacting cells with an amount of a composition
effective to inhibit the bacterial infection and/or to neutralize endotoxin,
wherein the composition includes one or more partial peptide mimetics
described herein.
In both the in vivo and in vitro methods, "inhibiting" a bacterial infection
includes preventing as well as reversing or reducing the growth of bacteria in
a
patient or a cellular sample, and "neutralizing" endotoxin includes binding
LPS
and thereby removing it from the system of a patient or a cellular sample. The
level of bacterial infection can be determined according to known bactericidal
assays. The level of endotoxemia can be determined according to known LPS
neutralization assays. These assays can be used to determine the effectiveness
of a polypeptide, whether used in vivo or in vitro. To determine the
effectiveness of the treatment of a patient having a bacterial infection, a
blood
sample can be taken, a culture developed, and the amount of live bacteria
determined. To determine the effectiveness of the treatment of a patient
having
endotoxemia, a blood sample can be taken, a culture developed, and the amount
of cytokines (e.g., TNF-a, IL-1) can be determined using methods known to one
of skill in the art. For example, the WEHI assay can be used for the detection
of
TNF-a (Battafarano et al., Surgery 118, 318-324 (1995)).
The effective amount of a partial peptide mimetic of the present
invention will depend on the condition being treated and on the desired
result.
For example, treating a bacterial infection will depend on the bacterial
infection,
the location of the infection, and the partial peptide mimetic. An effective
amount of the partial peptide mimetic for treating bacterial infection is that
amount that diminishes the number of bacteria in the animal and that
diminishes
19

CA 02476427 2004-08-16
WO 03/070751 PCT/US03/05106
the symptoms associated with bacterial infection such as fever, pain, and
other
associated symptoms of the bacterial infection. The effective amount of a
peptide can be determined by standard dose response methods.
Alternatively, an effective amount of a partial peptide mimetic for
treating a bacterial infection can be determined in an animal system such as a
mouse. Acute peritonitis can be induced in mice such as outbred Swiss webster
mice by intraperitoneal injection with bacteria such as P. aeruginosa as
described by Dunn et al. (Surgery, 98:283, 1985); Cody et al. (Int. Surg.
Res.,
52:315, 1992). Bactericidal activity can be evaluated against a variety of
bacteria, preferably Gram-negative bacteria, but the types of bacteria can
include
Pseudomonas spp including P. aeruginosa and P. cepacia, E. coli strains,
including E. coli B, Salmonella, Proteus mirabilis and Staphylococcus strains
such as Staphylococcus aureus. Partial peptide mimetics with endotoxin
neutralizing activity can be used to treat mammals infected with Gram-negative
bacteria systemically and that exhibit symptoms of endotoxin shock such as
fever, shock, and TNF-a release.
Endotoxin neutralizing activity can be measured by determining the
molar concentration at which the peptide completely inhibits the action of
lipopolysaccharide in an assay such as the Limulus amoebocyte lysate assay
(LAL, Sigma Chemicals, St. Louis, MO) or the chromogenic LAL 1000 test
(Biowhittacker, Walkersville, MD). Endotoxin neutralizing activity can also be
measured by calculating an inhibitory dose 50 (LDSO) using standard dose
response methods. An inhibitory dose 50 is that amount of peptide that can
inhibit 50% of the activity of endotoxin.
The present invention also provides a method for inhibiting the amount
of TNF-a in a patient (e.g., a mammal such as a human). This involves
administering to a patient an amount of a composition effective to inhibit the
amount of TNF-a in a patient's system as determined by evaluating serum
levels of TNF-a, wherein the composition includes one or more partial peptide
mimetics described herein. Analogously, the present invention provides a
method for inhibiting the amount of TNF-a in vitro (e.g., in a cell culture).
This
method involves incubating cells with an amount of a composition effective to

CA 02476427 2004-08-16
WO 03/070751 PCT/US03/05106
decrease TNF-a amounts in the cell culture, wherein the composition includes
one or more partial peptide mimetics described herein. For both in vivo and in
vitro methods, the WEHI assay can be used for the detection of TNF-a
(Battafarano et al., Surgery, 118, 318-324 (1995)) in cell culture or in serum
from a patient. Alternatively, the amount of TNF-a in a sample can be assayed
using an anti-TNF-a antibody. A partial peptide mimetic "active" for
decreasing TNF-a can be evaluated using an in vitro test, and preferably shows
an at least 10% decrease in the amount of TNF-a.
The invention will be further described by reference to the following
detailed examples. These examples are offered to further illustrate the
various
specific and preferred embodiments and techniques. It should be understood,
however, that many variations and modifications may be made while remaining
within the scope of the present invention.
Examples
EXPERIMENTAL PROTOCOL
Peptide Synthesis.
Peptides were synthesized using a Milligen/Biosearch 9600 peptide
solid-phase synthesizer using fluorenylmethoxycarbonyl chemistry. Lyophilized
crude peptides were purified by preparative reversed-phase HPLC on a C 18
column with an elution gradient of 0-60% acetonitrile with 0.1 %
trifluoroacetic
acid in water. The purity and composition of the peptides were verified by
HPLC (Beckman Model 6300) analysis of amino acid composition of
hydrolysates prepared by treating the peptides under argon in 6 Normal (6 N)
HCl for 24 hours at 110°C. The amino acid sequences of peptides
were
confirmed by N-terminal sequencing and mass spectrometry.
Synthesis of DBF Analogs.
Unexceptional phases of solid-phase peptide synthesis (SPPS) were
carried out on a Milligen/Biosearch 9600 peptide synthesizer using
fluorenylmethoxy-carbonyl (Fmoc) methodology and BOP/HOBT as coupling
reagents. Fmoc-DBF-COzH (1) was prepared by slight modification of the
21

CA 02476427 2004-08-16
WO 03/070751 PCT/US03/05106
reported method (H. Bekele et al., J. Org. Chem., 62, 2259-2262 (1997).). In
this nine-step synthesis, the intermediates and final product were
characterized
by TLC and 'H NMR, with GC-LRMS, "C NMR, melting points (mp), and IR
being used as appropriate. Coupling of 1 to ho (isoleucine at residue 20 of
the
~3pep-25 sequence) as well as of L" (leucine at residue 11 of the ~3pep-25
sequence) to the peptide-DBF-NHZ was performed on the synthesizer. Coupling
of Fmoc-K,o COzH (wherein K,o refers to the lysine at residue 10 of the (3pep-
25
sequence) to the peptide-DBF-L"-NHZ sequence was difficult and required
manual SPPS using the more reactive HATU reagent (L.A. Carpino, J. Am.
Chem. Soc., I15, 4397-4398 (1993)).
The remaining chemical couplings used for production of the DBF
analogs listed in Table 1 were carried out using BOP/HOBT conditions on the
peptide synthesizer. After the final Fmoc deprotection, each of the DBF
peptide analogs (partial peptide mimetics) was released from the resin with
simultaneous removal of all acidolyzable trityl and tert-butyl side-chain
protecting groups using Reagent K (D.S. King et al., Int. J. Pept. Protein.
Res.,
36, 255-266 (1990)). A Rink amide or similar resin was used to provide the
primary amide form of the C-terminal D24 unit. Lyophilized crude peptides
were purified by preparative reversed-phase HPLC on a C18 column using a
Hewlett-Packard 1090 system. An elution gradient of 0-60% acetonitrile in
water (0.1 % trifluoroacetic acid) was used. The purity and composition of the
peptides was verified by analytical HPLC, matrix assisted laser desorption
ionization (MALDI) mass spectrometry using a Hewlett-Packard G2025A
system and sinapinic acid as matrix, and analysis of amino acid composition of
hydrolysates (6N HCI, 110°C, 24 hours (h), under argon).
NMR Spectroscopy.
For NMR measurements, freeze-dried DBF analogs were dissolved in
water-DMSO-DPC mixture. Peptide concentration was 3 mM. pH was
adjusted to pH 5.7 by adding p.L quantities of NaOD or HCl to the peptide
sample. NMR spectra were acquired on a Varian UNITY Plus-600 NMR
spectrometer.
22

CA 02476427 2004-08-16
WO 03/070751 PCT/US03/05106
2D-homonuclear TOCSY with DIPSI (M. Piotto et al., J. Biomol. NMR,
2, 661-665 (1992)) spinlock (mixing time 80 milliseconds (ms)) was used to
identify spin systems. 2D NOESY experiments (G. Wider et al., J. Magn.
Reson., 56, 207-234 (1985)) were performed for sequential assignments and
conformational analysis. WATERGATE (A.J. Shaka et al., J. Magn. Reson., 77,
274-293 (1988); and S.P. Rucker et al., Mol. Phys., 68, 509-517 (1989)) was
used to attenuate the water resonance. All spectra were collected at
25°C as 256
to 512 tl experiments, each with 2048 complex data points over a spectral
width
of 6 kHz in both dimensions with the carrier placed on the water resonance.
Sixteen scans were time averaged per tl experiment. Data were processed
directly on the spectrometer using VNMR (Varian, Inc., Palo Alto) or NMRPipe
(F. Delaglio et al., J. Biomol. NMR, 6, 277-293 (1995)) on an SGI workstation.
Structural Modeling.
Analysis of NOE growth curves indicated that backbone to backbone
inter-proton NOES were normally maximum at about 200 ms. Interproton
distance constraints were derived from NOES assigned in'H NOESY spectra
acquired with mixing times of 100 ms. NOEs were classified as strong,
medium, weak, or very weak corresponding to upper bound distance constraints
of 2.8, 3.3, 4.0, and 4.5 ~, respectively. The lower bound restraint between
non-bonded protons was set to 1.8 ~I. Pseudo-atom corrections were added to
the upper bound distance constraints where appropriate, and a 0.5 ~ correction
was added to the upper bound for NOEs involving methyl protons. Hydrogen
bond constraints were identified from the pattern of sequential and
interstrand
NOEs involving NH and CaH protons, together with evidence of slow amide
proton-solvent exchange. Each hydrogen bond identified was defined using two
distance constraints; rNH-o = 1.8 to 2.5 ~, and rN-o = 1.8 to 2.5 ~.
Derived internuclear distance constraints were used in calculating
structures for 6DBF7 by using X-PLOR (A.T. Brunger, X plor Manual, Yale
University Press, New Haven (1992)). The molecule was created using
parallhdg.pro force fields. A template coordinate set was generated by using
the
Template routine. The ab initio simulated annealing (SA) protocol was then
23

CA 02476427 2004-08-16
WO 03/070751 PCT/US03/05106
used. The SA procedure ran high temperature dynamics (3000 K for 120
picoseconds (ps)) and then cooled down to 100 K in 50 K steps with 1.5 ps
molecular dynamics at each step. Powell minimization was performed at 100 K
for 1000 steps. Structure refinement was done based on simulated annealing
starting at 1000 K and ending at 100 K. Final structures were subjected to the
X-PLOR Accept routine with the violation threshold for NOES of O.St~ and
dihedral angles of 5°. Angles, bond lengths or impropers were not
allowed to
deviate from ideal geometry more than 5°, 0.05 A and 5°,
respectively.
Structures were superimposed using the BIOSYM INSIGHT viewer (Molecular
Simulations, Inc.) and were analyzed using X-PLOR analysis routines.
Cells, Cultures, and Reagents.
Human umbilical vein derived EC (HUVEC) were harvested from
normal human umbilical cords by perfusion with 0.125% trypsin/EDTA.
Harvested HUVECs were cultured in gelatin coated tissue culture flasks and
subcultured 1: 3 once a week in culture medium (RPMI-1640 with 20% human
serum (HS), supplemented with 2 mM glutamine and 100 U/ml penicillin and
0,1 mg/ml streptomycin). Bovine capillary EC (BCE) were kindly provided by
Dr. M. Furie (State University of New York, Stony Brook, USA) and were
cultured in fibronectin coated tissue culture flasks in RPMI-1640 medium
containing 10% FCS, glutamine and antibiotics.
EC Proliferation Measurement.
EC proliferation was measured using a [3H]- thymidine incorporation
assay. Proliferation of bFGF-stimulated (10 nanogram per milliliter (ng/ml))
human umbilical vascular EC (HUVEC) cultures was measured by
quantification of 3H-thymidine incorporation. Proliferation is expressed as
mean
counts per minute (cpm) of quadruplicate cultures in three independent
experiments (~ SEM). EC were seeded at 5000 cells/well in flat-bottomed
tissue culture plates and grown for 3 days in the absence or presence of
regulators, in culture medium. During the last 6 hours of the assay, the
culture
was pulsed with 0.5 ~Ci [methyl 3H]- thymidine/well. Human umbilical vein
24

CA 02476427 2004-08-16
WO 03/070751 PCT/US03/05106
derived EC (HUVEC) were harvested from normal human umbilical cords by
perfusion with 0.125% trypsin/EDTA. Harvested HUVECs were cultured in
gelatin coated tissue culture flasks and subcultured 1: 3 once a week in
culture
medium (RPMI-1640 with 20% human serum (HS), supplemented with 2 mM
glutamine and 100 U/ml penicillin and 0.1 mg/ml streptomycin).
In vitro Angiogenesis Assay.
Sprouting and tube formation of bovine EC (BCE) were studied using
cytodex-3 beads overgrown with BCE in a 3-dimensional collagen gel
(vitrogen-100, Collagen Corp., Fremont, CA, USA) as described in D. Van der
Schaft et al., Blood, 96, 176-181 (2000). Following gelation, culture medium
containing 20 ng/mL bFGF, with or without ~3pep-25 or DBF analogs, was
applied on top of the gel. After 24 hours of cell culture at 37°C,
photographs
were made (not shown). The amount of sprouting in each well (i.e., the total
1 S length of the sprouts) was quantified by the computer program NIH image.
To
quantify differences in sprouting and tube formation, statistical analysis was
performed using the Mann-Whitney U test. Using these data the ICSO on EC
proliferation and tube formation were quantified and are listed in Table 1.
Tumor Model Studies.
In all studies, female athymic nude mice (nu/nu, 5-6 weeks old) were
used. These mice were purchased from the National Cancer Institute and
allowed to acclimatize to local conditions for at least one week. Animals were
given water and standard chow ad libitum, and were kept on a 12-hour
light/dark cycle. All experiments were approved by the University of Minnesota
Research Animal Resources ethical committee. Mice were randomized and
split into three groups: 1) human serum albumin (10 mg/kg/day), 2) (3pep-25
(10
mg/kg/day) and 3) DBF analog (10 mg/kg/day). Compounds were diluted in
100 mM SDS and administered using osmotic mini-pumps (Durect, Cupertino,
CA). Exponentially growing MA148 human ovarian carcinoma cells, kindly
provided by Prof. Ramakrishnan (R.P. Dings et al., Cancer Res., 63, 382-385
(2003)) were cultured in RPMI 1640 medium (Life Technologies, Grand Island,

CA 02476427 2004-08-16
WO 03/070751 PCT/US03/05106
NY). This medium was supplemented with 10% fetal bovine serum and 1
penicillin/streptomycin (Cellgro, Mediatech, Washington, DC) at 37°C
and 5%
C02. One hundred microliters (100 ~L) of this tumor cell suspension (2 x 10'
cells/ml) was then injected subcutaneously into the right flank of each mouse.
S Pumps were implanted into the left flank of mice for subcutaneous
administration of compound over a 28-day treatment span.
Two variants of this model were used: prevention and intervention. For
the prevention variant, treatment was initiated at the time of inoculation
with
MA148 cells. For the intervention variant, tumors were allowed to grow to an
average size of 50 mm3 (usually day 7 post inoculation) before treatment was
initiated. With either variant, animals were randomized prior to the
initiation of
treatment. Treatment was administered via osmotic mini-pumps (Durect,
Cupertino, CA), which were implanted subcutaneously in the left flank of mice.
Concentrated solutions of (3pep-25 or DBF analogs were formulated such that
the 28-day treatment period would be covered by implantation of a single pump.
In each study, control groups of animals were administered either PBS or PBS
containing human serum albumin. Tumor growth curves were found to be
virtually identical in either of these control cases.
Tumor volume was determined by measuring the diameters of tumors
using calipers (Scienceware, Pequannock, NJ) using the equation for the volume
of a spheroid: (a2 x b x II) / 6, where 'a' is the width and 'b' the length of
the
tumor. Measurements were performed two or three times per week. At the
conclusion of an experiment, tumor weights were also taken following excision
of the tumors from euthanized animals. Tumor weights correlated well with
tumor volumes calculated in this way.
Immunohistochemistry.
Immunohistochemistry was used to assess microvessel density and the
extent of total cell apoptosis. Tumor tissue was embedded in tissue freezing
medium (Miles Inc, Elkart, IN) and shock frozen in liquid nitrogen. Ten
millimeter (10 mm) thick sections of tissue were prepared for
immunohistochemical analysis. For this, tissue sections were brought to room
26

CA 02476427 2004-08-16
WO 03/070751 PCT/US03/05106
temperature, air dried overnight, and then fixed in acetone for 10 minutes.
Slides were allowed to air dry for at least 30 minutes and were washed three
times for 5 minutes each in phosphate-buffered saline (PBS, pH 7.4). Samples
were then blocked with PBS containing 0.1 % bovine serum albumin and 3%
human serum albumin for at least 30 minutes at room temperature in a
humidified box. Samples were subsequently incubated with phycoerytrin (PE)-
conjugated monoclonal antibody to CD31 (PECAM-1) in a 1:50 dilution
(Pharmigen, San Diego, CA) to stain for microvessel density. After 1-hour
incubation at room temperature, slides were washed with PBS and immediately
imaged using an Olympus BX-60 fluorescence microscope at 200X
magnification.
To assess the extent of total cell apoptosis, tissue sections were stained
by using the TUNEL (terminal deoxyribonucleotidyl transferase-mediated
dUTP-nick-end labeling) assay, which was performed according to the
manufacturer's instructions (in situ cell death detection kit, fluorescein;
TUNEL,
Roche). Digital images were stored and processed using Adobe Photoshop
(Adobe Inc., Mountain View CA).
Quantification of microvessel density, the rate of proliferation and total
cell apoptosis were determined as described earlier (R. Wild et al.,
Microvasc.
Res., 59, 368-376 (2000)). Statistical analysis was performed using the
Student's t test.
Toxicity Assays.
As an indirect measurement of general toxicity, body weights of mice
were monitored twice weekly, using a digital balance (Ohaus Florham, NJ). To
determine hematocrit and creatinine levels, blood samples were extracted by
tail
vein bleedings one day after terminating treatment and blood was collected in
heparinized micro-hematocrit capillary tubes (Fisher; Pittsburgh, PA). For
hematocrit levels, samples were spun down for 10 minutes in a micro-
hematocrit centrifuge (Clay-Adams; NY), and the amount of hematocrit was
determined using an international microcapillary reader (IEC; Needham, Mass).
To obtain creatinine levels, a kit was purchased from Sigma (Sigma
27

CA 02476427 2004-08-16
WO 03/070751 PCT/US03/05106
Diagnostics; St Louis, MO) and used according to the manufacturer's
instructions.
RESULTS AND DISCUSSION
Detailed Structure-Activity Evaluation.
For input into designing the partial peptide mimetics of the present
invention, a complete structure-activity analysis with (3pep-25 was performed.
Working with this relatively small peptide allowed for a thorough assessment
of
desirable residues using alanine scanning and walk-through variants.
Dodecapeptide walk-through variants that walk through the (3pep-25
sequence, shifting three residues in each peptide were prepared and are listed
in
Figure 1 A. Their effect on EC proliferation is also indicated in Figure 1 A
with
IC50 values. Only three walk-through peptides demonstrated any significant
anti- proliferative activity relative to [3pep-25, and, as with results from
alanine
scanning, these peptides also encompass ~3-strands l and 2. Based on these
results, it can be concluded that activity is localized within (3-strands 1
and 2.
For orientation, the (3-strand alignment for ~3pep-25 is depicted in Figure
1B,
with key sequences being boxed-in.
Residues that demonstrated the most significant drop in the ability of
~ipep-25 to inihibit EC proliferation are hydrophobic residues within the
first
two (3-strands: I3, LS, V~, L~,, and IZO (these refer to the specific amino
acid
residues at the position indicated by the subscript in (3pep-25).
Conformationally, these hydrophobic residues lie on the same face of the
amphipathic anti-parallel (3-sheet.
Using these structure-activity relationships, a series of new molecules
were designed. In these, the ~i-strand 3 and turn 2 of (3pep-25 were omitted
and
a dibenzofuran (DBF) (3-turn mimetic (H. Diaz et al., J. Am. Chem. Soc., ll S,
3790-3791 (1993); and K.Y. Tsang et al., J. Am. Chem. Soc., 116, 3988-4005
( 1994)) was used in place of turn l and the remainder of ~3-strand 1
(residues 1-
11 of (3-pep-25 or SEQ ID NO:1) and (3-strand 2 (residues 20-26 of ~3pep-25 or
28

CA 02476427 2004-08-16
WO 03/070751 PCT/US03/05106
SEQ ID NO:1 ). The DBF ~3-turn mimetic was used in order to maintain the
bioactive (3-sheet conformation of (3pep-25.
A number of (3-turn inducing scaffolds are known. The dibenzofuran
scaffold developed by Kelly et al. (H. Diaz et al., J. Am. Chem. Soc.,115,
3790-
3791 ( 1993); and K.Y. Tsang et al., J. Am. Chem. Soc., 116, 3988-4005 (
1994))
is an a, w-amino acid that has been incorporated into peptides of varying
length
by both the Kelly group and others. Notably, replacement of two, ~3-turn-
inducing, natural amino acids in the protein scyllatoxin (a 31-mer) by the DBF
scaffold provided an analog whose secondary structure and activity were
indistinguishable from the parent toxin. Kelly has demonstrated that when the
DBF unit is inserted between a pair of lipophilic amino acid residues, a
"hydrophobic cluster" is created wherein the sidechains of those residues are
nested within the hydrophobic pocket created by the aromatic rings of the
canted
DBF subunit. Because of the desire to design peptide analogs of relatively
small
size, DBF is an excellent choice since it could be inserted between residues
IZo
and L1 ~ of (3pep-25. The lypophilic isoleucine at residue 20 is one of the
important, hydrophobic residues associated with ~3pep-25 anti-angiogenic
activity.
The Fmoc-protected version of the Kelly DBF is shown below in
Scheme 1 with several subtle improvements made in the synthesis (bolded
conditions).
29

CA 02476427 2004-08-16
WO 03/070751 PCT/US03/05106
Scheme 1
1 ) H2, Pd/C, EtOH
/ ~ 3 steps 2) ICI, K2C03
\ ~ 3) Heck coupling
O 4) H2, PdIC
EtOZC
1) (Ph0)2P(O)N3, NEt3
2) toluene, 70° C
3) 9-Fluorenemethanol
4) HCI, AcOH
Et( '2H H02C N Hh moc
dibenwftu~an (DBF-Fmoc)
The parent DBF-based compound depicted in Figure 1 C is called
11DBF7, where numbers at the left and right of DBF refer to the number of
amino acid residues in the N- and C-terminal strands from (3pep-25,
respectively, with the DBF turn mimic replacing amino acid residues 12-19 of
(3pep-25, SEQ ID NO:1. To identify the shortest sequences required for
bioactivity, a series of N- and C-terminal deletion variants of 11 DBF7 were
made as listed in Table 1. In the provisional application to which this
application claims priority, some of these were referred to as CF-1 through CF-
8. Furthermore, in the provisional application to which this application
claims
1 S priority, the segments of SEQ ID NO: I were written in reverse order
(i.e., from
the C-terminus to the N-terminus), which upon reading the context of the
provisional application would be clear to one of skill in the art. For
example,
CF-1 was written as DN25LKVII[DBF]L, wherein the N25 indicates the
asparagine is residue 25 of (3pep-25 or SEQ 1D NO:1.

CA 02476427 2004-08-16
WO 03/070751 PCT/US03/05106
~ o
\° '~? \° \° °
° O l~ O N o O
V1 M
'"~ +~ +I +I +i +I +I +I
M (~ ~D O 01 ~ '
N ~n I~ ~O Ov o0
0
O
~n '~
U ~ N \ o a ~ o o °
N ~ N Q~ l~ 00 ~ ~ o
O~ ~O v~ ~O d O
'\
0
O
°~
N N O ~n ~ ~ ,
c~ W ~--~ M .~ .~ N ,..,.~ N
...,
N
O
U
tj N N N N N ~ j N M
.o a
o O O O o p o
z z z z z z z
zr~zr~z~zr~z~zr~zA
~ a~ a~ a~ a~ a~ a~ a
~~~~~~>~?~?~~~
w
_ w w w w w w w w ~,, w ~., ~., w w
~) f~ f.~ G4 f~ G4 ~C1 GA pq G4 ~q ~G1 ~1 G4
~~1~~lf~~l~~~Ca~~L~1
4-~ N O a ~ a r1 a ~ ' n ' n ' a a
o tn a z ~ oo ~ ~ ~ .o ~ ~, ~ ~ ~
~o~o~o~o~o~
z z z z z
w~ ~a~a~a~a a
w ~ w ~ w w w
': r~ ~ r~ ~ r~ tn v~
a z ~- z ~-
A ~ d
d
U
b
o t~ O ~ M N
w w w w w w w
z
' o ~' ~'r ~, ri ri w w r3 ri
°~ ø~ G4 GA L~ ~ GA GA
~ o ~' ~ ~ o
E-~ U e2 L1 ,~
~n o ~n o ~n
N N

CA 02476427 2004-08-16
WO 03/070751 PCT/US03/05106
A ~ ~ A ~ ~ A
z z z z z z z z
0
+i
+~ ~. , , ,
M N ' ' ' ~D
N M N ~' i i i l~
Cn M ~ M
M
N
z z z z
zr~ r~ z r~
~a ~a ~ a~a
r, " ' ,; ' ~., ' '
ww ww w w ,--, ' r, w ~
w ~
w w w ~
w ~
. .
r~r~ ~a~r.~ aa c~a. r~aa
.
~c~r~r~c~c~r.~
aa aaa a a a
a,
,a
a
n n ~ u " "
00 00 00 ' - tn
a
~ ' n n ' n
V~ ~ ~ V~
.~'',
az az z a z a z a z a a z
a z
>~ >r~> a > ~>r~>r~>r~ > A
~a ~a~' a ~ a'~a~a~a ~ a ~ A
.~ .~ .~ ~ .~ ~. ~, ~ o
z z
o n
o z
0
w
..,
o
v0 ~f~- U _
w ww w w w w w ''"'
0
z
o ~ o
N

CA 02476427 2004-08-16
WO 03/070751 PCT/US03/05106
(3-Sheet Structure is Preserved in DBF Analogs.
NMR spectroscopy was used to investigate whether ~3-sheet
conformation was preserved in DBF analogs. Analogs 11 DBF7 and 6DBF7 that
have more equivalent (3-strand lengths, were the focus of this structural
study.
Due to limited water solubility and the desire to mimic a membrane-like
environment, these compounds were investigated in dodecylphosphocholine
(DPC) micelles. At the millimolar concentrations required for NMR work,
11DBF7 gave overlapping resonances that made spectral analysis ambiguous.
6DBF7 gave excellent NMR spectra that allowed complete structure analysis.
NOES and coupling constants diagnostic of anti-parallel ~3-sheet conformation
were readily identified and used in computational modeling. Superposition of
the resulting 28 structures is shown in Figure 2A, along with a simplified
illustration of this folding pattern in Figure 2B, which highlights residues
on
both hydrophobic and hydrophilic surfaces of the (3-sheet. Notice in this
structure that the two aliphatic hydrophobic residues Leu and Ile in 6DBF7 are
packed against the phenyls of the DBF group. In effect, this sets up and helps
stabilize the (3-sheet fold. Based on this structural information on 6DBF7, it
can
be concluded that other DBF analogs would fold similarly, albeit to various
extents depending on the lengths of the two strands, i.e., strands of equal
length
are expected to be better able to form (3-sheets.
DBF Analogs Inhibit EC Proliferation and Retain Anti-angiogenic Activity.
In endothelial cell (EC) proliferation assays, it was demonstrated that
11DBF7, as well as a number of shorter analogs, are effective at inhibiting EC
growth. Dose response curves for all xDBF7 analogs are exemplified in Figure
3, and ICSO values for all DBF analogs (i.e., partial peptide mimetics) and
percent inhibition of EC growth at 25 ~M are given in Table 1. Compared to
parent (3-pep-25 (IC50, 3 ~.M), most DBF analogs, although slightly less
active
(IC50 about 10 ~.M), do perform relatively well in this assay. This may, in
part,
be explained by their decreased size. In addition, some of the analogs are
missing residues identified by alanine scanning as being functionally
important
33

CA 02476427 2004-08-16
WO 03/070751 PCT/US03/05106
(e.g., LS and I3). Nonetheless, a number of these shorter analogs remain
reasonably active, and it appears that the N-terminal hexapeptide SVQMKL
(SEQ ID NO:S) and C-terminal tetrapeptide IIVK (SEQ ID N0:3) enhance anti-
angiogenic activity.
S Angiostatic potential was further demonstrated in vitro in the collagen
gel-based sprout formation assay (R.P. Dings et al., Cancer Res., 63, 382-385
(2003)). Whereas the control showed numerous sprouts, treatment with (3pep-
25, 11 DBF7 and 6DBF7 all demonstrated highly reduced sprouting (data not
shown). Quantitative results for all analogs are given in Table 1. In general,
reducing the number of amino acid residues in the ~i-strands led to reduced
inhibition of sprout formation, comparable with the proliferation assay.
However, some analogs were active and even one of the shortest analogs,
6DBF7, had a significant inhibitory effect on tube formation. The kinetics of
inhibition using DBF analogs, moreover, were the same as those for (3pep-25
(data not shown).
DBF Analogs Inhibit Tumor Growth in Mice.
Because specific use of an anti-angiogenic agent is in the area of tumor
biology, the in vivo efficacy of two of the most in vitro active DBF analogs,
11DBF7 and 6DBF7, were assessed along with (3pep-25 as a positive control, in
the MA148 xenograft ovarian carcinoma tumor model in athymic mice as
previously described in R.P. Dings et al., Cancer Res., 63, 382-385 (2003).
Initial experiments using this model administered the DBF analog, 11DBF7,
subcutaneously via mini-pumps implanted at the time of inoculation with the
tumor cell line. This prevention model demonstrated that treatment of tumor
forming animals with 11DBF7 resulted in inhibition of tumor growth (Figure
4A). Surprisingly, 11DBF7 functioned, on average, slightly better than (3pep-
25
by reducing tumor volume by up to 90% relative to control animals.
In further experiments, treatment was initiated seven days following
inoculation with tumor cells to allow establishment of small tumors prior to
the
start of treatment. Using this protocol, (3pep-25, 11 DBF7, as well as 6DBF7,
34

CA 02476427 2004-08-16
WO 03/070751 PCT/US03/05106
were tested and were found to inhibit tumor growth by up to 70% during the
course of treatment and by about 50% at the end of the four-week
administration
period (day 35) (Figure 4B). The rate of tumor growth then began to increase,
but remained at about 50% ten days post-treatment when animals were
sacrificed for analysis of tumor tissue. Interestingly, the smaller DBF
analog,
6DBF7, was observed to be slightly more effective at inhibiting tumor growth
than (3pep-25 or 11 DBF7. On day 35, for example, 6DBF7 sustained tumor
growth inhibition to more than 70% relative to tumors from control animals
(Figure 4B).
In vivo anti-angiogenic potential was demonstrated by staining tumor
cross-sections from treated animals with fluorescently-labeled anti-CD31
antibody used immunohistochemically to identify blood vessels. Vessel density,
relative to control, was significantly reduced by treatment with ~3pep-25,
11 DBF7, or 6DBF7. In addition, these anti-angiogenic compounds had a
significant effect as well on vessel architecture, demonstrating a drop in the
number of end points, branch points and vessel length (Table 2). In addition,
anti-angiogenic treatment also reduced the rate of proliferation of tumor
cells as
determined by immunohistochemical staining of PCNA in cryosections of
tumors (Table 2). As a result of angiogenic inhibition, the number of
apoptotic
tumor cells increased from 311 t 103 in the control, to 620 ~ 146 and 851 ~
162
in ~3pep-25 and 6DBF7 treated animals, respectively.
Table 2.
Microvessel density and proliferation rate in tumors of treated mice
Proliferationa Vessel End Pointsc Branch Vessel
Densityb Pointsd Lengthe
Vehicle ~ 848 ~ 104 ~ 5858 ~ 656 26.2 ~ 2.2 7.6 ~ 1.4 5.9 ~ 0.7
~3pep-25 414 ~ 44 2245 ~ 329 22.9 ~ 2.2 2.1 ~ 0.6 0.9 ~ 0.3
11 DBF7 553 ~ 75 2879 ~ 385 21.2 ~ 3.3 3.1 ~ 0.9 3.0 ~ 0.6
6DBF7 ~ 501 ~ 68 I 2213 ~ 256 21.7 ~ 2.6 2.3 ~ 1.1 2.5 ~ 0.4

CA 02476427 2004-08-16
WO 03/070751 PCT/US03/05106
a After binarization of the images of the PCNA-staining, proliferation was
estimated by scoring the total number of white pixels per field.
b After binarization of the images of the CD31-staining, microvessel density
was
estimated by scoring the total number of white pixels per field.
'Mean number of vessel end points as determined after skeletonization of the
images (R. Wild et al., Microvasc. Res., 59, 368-376 (2000)).
Mean number of vessel branch points/nodes per image.
eMean total vessel length per image.
All results are expressed as mean pixel counts per image (~ standard error).
In all in vivo experiments, treatment with (3pep-25, 11 DBF7, and 6DBF7
did not show any sign of toxicity as assessed by unaltered behavior and normal
weight gain during experiments (data not shown). Moreover, hematocrit and
creatinine levels in treated animals were normal relative to control,
indicating
the absence of toxicity to bone marrow and kidney. Macro- and microscopic
morphology of internal organs on autopsy were also observed to be normal
within all experimental groups of animals.
Conclusion
In summary, amino acid residues that promote ~3pep-25's angiostatic
activity have been identified, and partial peptide mimetics of (3pep-25 have
been
designed and synthesized. The more effective mimetics carried only 13 of its
33
amino acid residues and showed that the partial peptide mimetic is an
effective
anti- angiogenic agent both in vitro and in vivo. Moreover, whereas as in
vitro
activity is somewhat less than that of (ipep-25, in vivo activity appears to
be
improved, suggesting increased bioavailabilty with DBF analogs. Both the
significant reduction in the number of residues required to promote
angiostatic
activity and the use of an organic (3-sheet-inducing scaffold allows for the
development of an orally-active, small molecule mimetic of an anti-angiogenic
protein.
36

CA 02476427 2004-08-16
WO 03/070751 PCT/US03/05106
The DBF analogs not tested in these analyses would be expected by one
of skill in the art to provide some activity in at least one of the assays due
to the
homology of their structures. Also, although not shown, DBF analogs of the
type described herein demonstrate some bacteriocidal activity and some
endotoxin neutralizing activity. Furthermore, because of the results shown
herein, DBF analogs of the type described herein would be expected by one of
skill in the art to inhibit TNF-a levels, promote inter-cellular adhesion
molecule
(ICAM) expression, and inhibit inter-cellular adhesion molecule (ICAM)
expression down regulation. Also, particularly because of the results with
respect to the inhibition of angiogenesis, DBF analogs of the type described
herein would be expected by one of skill in the art to inhibit pathologic
disorders such as atherosclerosis, restenosis, diabetic retinopathy,
neovascular
glaucoma, rheumatoid arthritis, and endometriosis.
All references cited herein are incorporated by reference, in their
entirety, into this text. Although the invention has been described in the
context
of particular embodiments, it is intended that the scope of coverage of the
patent
be limited only by reference to the following claims.
37

CA 02476427 2004-08-16
WO 03/070751 PCT/US03/05106
Sequence Listing Free Text
Amino Acid Sequence of Peptides
~ipep-25 (SEQ ID NO:1 )
S ANIKLSVQMKLFKRHLKWKIIVKLNDGRELSLD
(SEQ ID N0:2)
IIVKLND
(SEQ ID N0:3)
IIVK
(SEQ ID N0:4)
QMKL
(SEQ ID NO:S)
SVQMKL
(SEQ ID N0:6)
IKLSVQMKL
(SEQ ID N0:7)
NIKLSVQMKL
(SEQ ID N0:8)
ANIKLSVQMKL
(SEQ ID N0:9)
IIVKLN
38

Representative Drawing

Sorry, the representative drawing for patent document number 2476427 was not found.

Administrative Status

2024-08-01:As part of the Next Generation Patents (NGP) transition, the Canadian Patents Database (CPD) now contains a more detailed Event History, which replicates the Event Log of our new back-office solution.

Please note that "Inactive:" events refers to events no longer in use in our new back-office solution.

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Event History , Maintenance Fee  and Payment History  should be consulted.

Event History

Description Date
Deemed Abandoned - Failure to Respond to Maintenance Fee Notice 2009-02-20
Application Not Reinstated by Deadline 2009-02-20
Inactive: Dead - RFE never made 2009-02-20
Inactive: Abandon-RFE+Late fee unpaid-Correspondence sent 2008-02-20
Inactive: IPC from MCD 2006-03-12
Inactive: IPC from MCD 2006-03-12
Inactive: IPC from MCD 2006-03-12
Inactive: Sequence listing - Amendment 2005-08-12
Inactive: Office letter 2005-04-05
Inactive: IPC assigned 2005-01-31
Inactive: IPC assigned 2005-01-31
Inactive: IPC assigned 2005-01-31
Inactive: First IPC assigned 2005-01-31
Inactive: Cover page published 2004-12-13
Letter Sent 2004-12-10
Letter Sent 2004-12-10
Inactive: Notice - National entry - No RFE 2004-12-10
Inactive: IPC assigned 2004-10-28
Inactive: IPC assigned 2004-10-28
Inactive: First IPC assigned 2004-10-28
Inactive: IPC assigned 2004-10-28
Inactive: IPC assigned 2004-10-28
Application Received - PCT 2004-09-14
National Entry Requirements Determined Compliant 2004-08-16
Application Published (Open to Public Inspection) 2003-08-28

Abandonment History

Abandonment Date Reason Reinstatement Date
2009-02-20

Maintenance Fee

The last payment was received on 2008-01-31

Note : If the full payment has not been received on or before the date indicated, a further fee may be required which may be one of the following

  • the reinstatement fee;
  • the late payment fee; or
  • additional fee to reverse deemed expiry.

Please refer to the CIPO Patent Fees web page to see all current fee amounts.

Fee History

Fee Type Anniversary Year Due Date Paid Date
Basic national fee - standard 2004-08-16
Registration of a document 2004-08-16
MF (application, 2nd anniv.) - standard 02 2005-02-21 2005-02-01
MF (application, 3rd anniv.) - standard 03 2006-02-20 2006-01-31
MF (application, 4th anniv.) - standard 04 2007-02-20 2007-01-31
MF (application, 5th anniv.) - standard 05 2008-02-20 2008-01-31
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
REGENTS OF THE UNIVERSITY OF MINNESOTA
Past Owners on Record
CAROLEE FLADER LAVEY
KEVIN H. MAYO
THOMAS R. HOYE
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Description 2004-08-16 38 1,723
Claims 2004-08-16 8 251
Abstract 2004-08-16 1 53
Drawings 2004-08-16 4 104
Cover Page 2004-12-13 1 30
Description 2005-08-12 42 1,823
Reminder of maintenance fee due 2004-12-13 1 110
Notice of National Entry 2004-12-10 1 193
Courtesy - Certificate of registration (related document(s)) 2004-12-10 1 106
Courtesy - Certificate of registration (related document(s)) 2004-12-10 1 106
Reminder - Request for Examination 2007-10-23 1 119
Courtesy - Abandonment Letter (Request for Examination) 2008-05-14 1 167
Courtesy - Abandonment Letter (Maintenance Fee) 2009-04-20 1 172
PCT 2004-08-16 2 93
Correspondence 2005-04-05 2 31

Biological Sequence Listings

Choose a BSL submission then click the "Download BSL" button to download the file.

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.

Please note that files with extensions .pep and .seq that were created by CIPO as working files might be incomplete and are not to be considered official communication.

BSL Files

To view selected files, please enter reCAPTCHA code :